11 research outputs found

    Duplin expression does not rescue shChd8 phenotype in B-ALL cells.

    No full text
    <p><b>(A)</b> Western blot showing Duplin expression in B-ALL cells transduced with <i>MIG-Duplin</i>, but not untransduced cells. <b>(B)</b> Bar graph showing <i>Chd8</i> C<sub>T</sub> values in the indicated mouse tissues with the indicated qPCR primer pairs. Shown are averages ± SD. <b>(C)</b> Bar graph showing depletion of sh<i>Chd8</i>-expressing B-ALL cells transduced with <i>MIG-Duplin</i> or a GFP vector control. Shown are averages normalized to vector control ± SD. Fold changes were calculated from day 2 to day 10 after second retroviral infection and normalized to MLT (tdTomato) empty vector control.</p

    T cell malignancies exhibit different requirements for CHD8 expression that can be partially alleviated by constitutive Notch signaling.

    No full text
    <p><b>(A)</b> (Left) Graph showing results of <i>in vitro</i> growth competition assays with Top Notch cells. Shown are averages ± SEM of four independent experiments. (Right) Western blot showing decreased CHD8 expression in Top Notch cells following transduction with sh<i>Chd8</i>-0 or -1. <b>(B)</b> (Left) Graph showing results of <i>in vitro</i> growth competition assays with KP lymphoma cells. Shown are averages ± SEM of five independent experiments. (Right) Western blot showing decreased CHD8 expression in KP lymphoma cells following transduction with sh<i>Chd8</i>-0 or -1. Fold changes of competition assays were calculated from day 2 to day 10 after retroviral infection and normalized to an empty vector control. <b>(C)</b> Graph showing results of ICN rescue experiment with KP lymphoma cells. Shown are averages ± SD from a representative experiment. Fold changes were calculated from day 2 to day 10 after second retroviral infection and normalized to MLT empty vector control. *P < 0.05, ***P < 0.0005, ****P < 0.0001</p

    CHD8 knockdown in B-ALL cells leads to cell death without a cell cycle arrest.

    No full text
    <p><b>(A)</b> Flow cytometry plots showing induction of dsRed and a linked shRNA upon treatment with doxycycline. <b>(B)</b> Western blot showing decreased CHD8 expression over time following induction of sh<i>Chd8</i> in B-ALL cells by doxycycline. <b>(C)</b> Bar graph showing increased <i>CCNE1</i> and <i>CCNE2</i> expression upon CHD8 knockdown. Shown are averages ± SD. <b>(D)</b> Bar graph showing cell cycle profiles of B-ALL cells following shRNA induction by doxycycline. <b>(E)</b> Graph showing growth rates of B-ALL cells transduced with the indicated constructs. Shown are averages ± SD. <b>(F)</b> Bar graph showing percentages of dead cells following <i>Chd8</i> knockdown. Survival of cells transduced with sh<i>Chd8</i> may be due to selection against CHD8 knockdown. Shown are averages ± SD. *P < 0.01, **P < 0.005, ***P < 0.0005</p

    CRISPR-Cas9 editing of CHD8 is detrimental to B-ALL cells.

    No full text
    <p><b>(A)</b> Western blot showing loss of CHD8 expression or expression of a truncated protein following gene editing by CRISPR-Cas9 in B-ALL cells. Expression of truncated protein likely arises from an ORF in original reading frame following a new stop codon generated by gene editing. Chd8 sgRNA-1 and -2 cell lines were generated with sgChd8-2 construct, Chd8 sgRNA-4 was generated with sgChd8-3 construct, and Chd8 sgRNA-3 and -5 were generated with sgChd8-4 construct. <b>(B)</b> (Left) Bar graph showing results of <i>in vitro</i> growth competition assays with B-ALL cells following gene editing by CRISPR-Cas9. Constructs targeting Renilla luciferase (Ren) were used as negative controls. Ren-1 and -2 cell lines were generated using sgRen-2 construct, and Ren-3 was generated using sgRen-3 construct. Shown are averages ± SD. (Right) Detailed bar graph showing results of <i>in vitro</i> growth competition assays. <b>(C)</b> Kaplan-Meier curve showing survival of mice injected with B-ALL cells transduced with the indicated constructs. n = 10 per cohort. See <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0143275#pone.0143275.s013" target="_blank">S6 Table</a> for statistics. ****P < 0.0001</p

    CHD8 depletion is detrimental in multiple B cell malignancies and normal pre-B cells.

    No full text
    <p><b>(A)</b> Western blot showing CHD8 expression in B-ALL cells and untransformed pre-B cells. <b>(B)</b> Graph showing results of growth competition assay with pre-B cells transduced with the indicated constructs. Shown are averages ± SD. Fold changes were calculated from day 2 to day 10 after retroviral infection and normalized to an empty vector control. <b>(C)</b> Western blot showing decreased CHD8 expression following transduction of Eμ-myc <i>Arf</i><sup><i>-/-</i></sup> cells with the indicated sh<i>Chd8</i> constructs. <b>(D)</b> Graph showing results of <i>in vitro</i> growth competition assays with Eμ-myc cells transduced with the indicated constructs. Shown are averages ± SEM of four independent experiments. Fold changes were calculated from day 2 to day 10 after retroviral infection and normalized to an empty vector control. <b>(E)</b> Growth curves of Eμ-myc cells transduced with the indicated constructs. Shown are averages ± SD. <b>(F)</b> Bar graph showing percentages of dead cells following <i>Chd8</i> knockdown in Eμ-myc cells. Shown are averages ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001</p

    Summary of the characteristics, expression, the availability of mouse model, and association to cancers of B- and Y-family translesion synthesis polymerases.

    No full text
    <p>Summary of the characteristics, expression, the availability of mouse model, and association to cancers of B- and Y-family translesion synthesis polymerases.</p

    DNA damage bypass process.

    No full text
    <p>(A) Mechanism of the 2-step DNA damage bypass process. To bypass DNA damage, REV1 inserts deoxycytidine triphosphates across the damage or orchestrates the recruitment of the other polymerases, POL ι, POL κ, POL η, to replicate across the damage. Thereafter, POL ζ complex can help extend beyond the damage to enable re-initiation of undamaged DNA replication. If an incorrect nucleotide gets incorporated across the damage, this misincorporated nucleotide will lead to a mutation in the next round of replication. (B) A schematic representing the protein domains of the Y-family translesion synthesis (TLS) polymerases, REV1, POL ι, POL κ, POL η.</p

    Correction to “A Pt(IV) Pro-drug Preferentially Targets Indoleamine-2,3-dioxygenase, Providing Enhanced Ovarian Cancer Immuno-Chemotherapy”

    No full text
    Correction to “A Pt(IV) Pro-drug Preferentially Targets Indoleamine-2,3-dioxygenase, Providing Enhanced Ovarian Cancer Immuno-Chemotherapy

    Bidentate Ligands on Osmium(VI) Nitrido Complexes Control Intracellular Targeting and Cell Death Pathways

    No full text
    The cellular response evoked by antiproliferating osmium­(VI) nitrido compounds of general formula OsN­(N^N)­Cl<sub>3</sub> (N^N = 2,2′-bipyridine <b>1</b>, 1,10-phenanthroline <b>2</b>, 3,4,7,8-tetramethyl-1,10-phenanthroline <b>3</b>, or 4,7-diphenyl-1,10-phenanthroline <b>4</b>) can be tuned by subtle ligand modifications. Complex <b>2</b> induces DNA damage, resulting in activation of the p53 pathway, cell cycle arrest at the G2/M phase, and caspase-dependent apoptotic cell death. In contrast, <b>4</b> evokes endoplasmic reticulum (ER) stress leading to the upregulation of proteins of the unfolded protein response pathway, increase in ER size, and p53-independent apoptotic cell death. To the best of our knowledge, <b>4</b> is the first osmium compound to induce ER stress in cancer cells

    Bidentate Ligands on Osmium(VI) Nitrido Complexes Control Intracellular Targeting and Cell Death Pathways

    No full text
    The cellular response evoked by antiproliferating osmium­(VI) nitrido compounds of general formula OsN­(N^N)­Cl<sub>3</sub> (N^N = 2,2′-bipyridine <b>1</b>, 1,10-phenanthroline <b>2</b>, 3,4,7,8-tetramethyl-1,10-phenanthroline <b>3</b>, or 4,7-diphenyl-1,10-phenanthroline <b>4</b>) can be tuned by subtle ligand modifications. Complex <b>2</b> induces DNA damage, resulting in activation of the p53 pathway, cell cycle arrest at the G2/M phase, and caspase-dependent apoptotic cell death. In contrast, <b>4</b> evokes endoplasmic reticulum (ER) stress leading to the upregulation of proteins of the unfolded protein response pathway, increase in ER size, and p53-independent apoptotic cell death. To the best of our knowledge, <b>4</b> is the first osmium compound to induce ER stress in cancer cells
    corecore